Abstract
Advancements in messenger RNA (mRNA) technology have fundamentally reshaped modern vaccinology, providing an agile and scalable platform capable of rapid vaccine development. This review examines how innovations originally accelerated during the COVID-19 pandemic are now being extended to other major respiratory viruses, particularly influenza and respiratory syncytial virus (RSV). The purpose of the study is to evaluate the scientific foundations, clinical progress, and limitations of applying mRNA platforms beyond SARS-CoV-2. Relevant literature published between 2008 and 2024 was identified through PubMed, Scopus, and Web of Science using keyword combinations including “mRNA vaccines,” “influenza,” “RSV,” “clinical trial,” and “immunogenicity.” Peer-reviewed primary studies, regulatory documents, and systematic reviews were included, while commentaries and non-peer-reviewed sources were excluded unless confirmed by primary data. The findings indicate that mRNA vaccines offer multiple advantages over traditional platforms, including rapid sequence-based design, strong humoral and cellular immune responses, and the capacity to incorporate multivalent or conserved antigenic targets. Clinical data from candidates such as Moderna’s mRNA-1010 (influenza) and mRNA-1345 (RSV) show robust neutralizing antibody responses, durable immunity, and acceptable safety profiles. These outcomes highlight the potential for mRNA technology to overcome strain-mismatch issues in influenza vaccination and longstanding challenges in eliciting protective RSV immunity. Despite this promise, significant limitations remain. The need for ultra-cold storage restricts distribution in low-resource settings, and current LNP-based delivery systems introduce manufacturing complexity and reactogenicity concerns. Additionally, long-term safety and real-world effectiveness data for non-COVID-19 mRNA vaccines are still limited, underscoring the need for continued clinical monitoring and formulation improvement. Overall, mRNA technology represents a transformative tool for addressing viral diseases beyond COVID-19, yet further innovation in thermostability, delivery platforms, and global manufacturing capacity is essential for maximizing its future impact.
Keywords: mRNA vaccines; lipid nanoparticles (LNPs); nucleoside modification; self-amplifying mRNA (saRNA); SARS-CoV-2; COVID-19; influenza; respiratory syncytial virus (RSV); vaccine efficacy; immunogenicity; humoral and cellular immunity; Phase 3 clinical trials; cold chain and thermostability; manufacturing cost; vaccine equity; regulatory pathways
Introduction
The COVID-19 pandemic has exposed the vulnerability of global public health systems to emerging viral threats, disrupting lives and economies worldwide. By January 2025, SARS-CoV-2 caused over 700 million infections and millions of deaths globally, prompting an urgent search for rapid and effective vaccines1. Traditional vaccine platforms, including inactivated and live-attenuated vaccines, often require years of development and manufacturing, a timeline incompatible with the immediate demands of a global pandemic. This challenge catalyzed the rise of messenger RNA (mRNA) vaccine technology, which revolutionized vaccine development by significantly reducing production timelines and enhancing adaptability2.
mRNA vaccines represent a paradigm shift in vaccinology. Unlike conventional approaches, these vaccines use synthetic mRNA to instruct host cells to produce antigenic proteins, eliciting immune responses. The success of mRNA-based COVID-19 vaccines, such as those developed by Pfizer-BioNTech and Moderna, highlight their immense potential. Their rapid development, high efficacy, and scalable production provided a blueprint for combating future pandemics and overcoming long-standing challenges in vaccine development for diseases like influenza and respiratory syncytial virus (RSV). Specifically, Phase 3 trials of BNT162b2 and mRNA‑1273 reported ~95% and 94.1% efficacy against symptomatic COVID‑19, respectively3,4, achieved within a year of genome release—versus typical 10–15‑year timelines for conventional platforms.
Beyond COVID-19, the applicability of mRNA technology extends to other infectious diseases, including those that have eluded traditional vaccine strategies. Influenza, RSV, and even emerging zoonotic viruses could benefit from the modular and adaptable nature of mRNA platforms. The capability to rapidly design and produce vaccines based on genetic sequencing alone positions mRNA technology as a cornerstone of future pandemic preparedness and routine immunization programs.
This paper examines how advancements in mRNA technology have expanded its transformative influence beyond COVID-19. It first provides an overview of mRNA technology’s foundational principles and historical development. Next, it explores the role of mRNA technology in the creation of COVID-19 vaccines, emphasizing their adaptability and effectiveness. Finally, the discussion pivots to the broader application of mRNA vaccines in combating other viral diseases, emphasizing ongoing research and future directions. Influenza and RSV were prioritized because they impose major global burdens with inconsistent vaccine performance: influenza causes up to 41 million illnesses annually in the U.S. with average 40–60% vaccine effectiveness, and RSV remains a leading cause of hospitalization in infants and older adults5,6,7.
Methods
Literature was identified via PubMed, Web of Science, and Scopus using combinations of “mRNA vaccine,” “influenza,” “RSV,” “clinical trial,” and “immunogenicity,” covering 2008–2024. Inclusion: peer‑reviewed primary studies, systematic reviews, and regulatory documents reporting clinical or preclinical outcomes. Exclusion: non‑peer‑reviewed commentaries, press releases, and duplicates. Corporate data were corroborated with peer‑reviewed publications when available.
Background on mRNA Technology
Unlike conventional inactivated or protein‑subunit vaccines that predominantly elicit humoral responses, mRNA vaccines drive endogenous antigen production, enabling presentation via MHC I and II pathways to activate CD8⁺ and CD4⁺ T cells alongside B‑cell responses, contributing to breadth and durability8,9.
Messenger RNA (mRNA) technology leverages the natural role of mRNA in cellular processes to drive immune responses. mRNA serves as a transient intermediary between DNA and protein synthesis, carrying genetic instructions for cells to produce specific proteins. In the context of vaccines, synthetic mRNA encodes viral antigens, prompting the host’s immune system to recognize and neutralize future infections. Unlike traditional vaccines that rely on cultured pathogens or proteins, mRNA vaccines use chemically synthesized nucleotides, significantly accelerating the development process10,11.
The mRNA vaccine success relies on the stabilization of synthetic mRNA and efficient cell delivery. Early research faced significant hurdles, including mRNA’s inherent instability and its tendency to provoke aberrant immune responses. For instance, unmodified mRNA often triggered innate immune reactions, leading to inflammation and reducing the vaccine’s efficacy. In 2008, Karikó et al. demonstrated that chemical modifications to mRNA’s nucleosides—specifically incorporating pseudouridine—could reduce its immunogenicity while preserving its functionality, laying the groundwork for mRNA therapeutics12. These advancements improved the safety profile of mRNA-based interventions and enhanced their clinical viability.
The development of LNPs marked another major advancement in mRNA technology, providing a crucial delivery mechanism. LNPs protect mRNA from enzymatic degradation in the bloodstream and facilitate its entry into target cells. Upon cellular uptake, the mRNA is released into the cytoplasm, where it is translated into proteins that trigger immune responses11. Without such delivery systems, mRNA would rapidly degrade, rendering it ineffective as a therapeutic tool. Beyond non‑immunogenic nucleoside modification, self‑amplifying mRNA (saRNA) vectors incorporate alphaviral replicase to boost intracellular RNA copies, enabling lower doses, and modern LNPs employ ionizable lipids and helper components that enhance endosomal escape and biodistribution while reducing reactogenicity13,9.
The historical trajectory of mRNA technology reflects decades of iterative advancements. Initial studies in the 1990s highlighted the potential of mRNA as a therapeutic agent but revealed critical challenges related to its instability and delivery8. By the early 2000s, researchers had addressed these limitations through improved transcription methods, codon optimization, and enhanced purification techniques14. The advent of high-fidelity RNA polymerases and cap analogs further increased synthetic mRNA’s stability and translational efficiency10.
Building upon these advancements, one of the defining advantages of mRNA technology is its flexibility, which refers to its ability to rapidly generate and modify vaccine formulations without the need for extensive reengineering or complex production processes. Unlike traditional vaccines, which require cultivating pathogens or extracting proteins, mRNA vaccines allow researchers to adapt quickly to new pathogens by altering the genetic sequence of the mRNA payload11. This adaptability is particularly valuable for addressing rapidly mutating viruses such as influenza and SARS-CoV-2, where quick response times are crucial for effective disease control15. Furthermore, mRNA vaccines can incorporate multiple antigenic targets within a single formulation, enabling broader protection against diverse viral strains and emerging variants9.
Ongoing research continues to examine the safety and the side effect profiles of mRNA vaccines. While mRNA vaccines have demonstrated excellent safety records, researchers have observed rare adverse events, such as myocarditis, particularly in younger populations. Understanding the mechanisms underlying these side effects and refining formulations to mitigate them remain critical for ensuring public confidence in mRNA-based therapeutics10.
Beyond infectious diseases, mRNA technology offers promising applications in cancer immunotherapy and protein replacement therapies. Clinical trials are exploring mRNA vaccines targeting oncogenic antigens, leveraging the same principles used in infectious disease vaccines. Additionally, mRNA’s ability to encode therapeutic proteins offers potential treatments for genetic disorders and chronic diseases. As researchers refine mRNA-based therapies, they are expanding its application beyond vaccination to include regenerative medicine and targeted molecular therapies.
In summary, mRNA technology represents a groundbreaking innovation in medical science. Its modular design, rapid production capabilities, and adaptability to diverse applications have established it as a cornerstone of modern vaccinology and therapeutics. As researchers continue to address challenges related to stability, delivery, and safety, the potential of mRNA technology to revolutionize healthcare remains boundless.
Role of mRNA Technology in COVID-19 Vaccine Development
The Emergence of COVID-19 and the Revolutionary Approach
COVID-19, caused by the novel coronavirus SARS-CoV-2, emerged in late 2019, triggering a global health crisis. Characterized by respiratory symptoms, systemic inflammation, and high transmission rates, the virus necessitated a rapid and effective vaccine strategy. mRNA technology emerged as a frontrunner due to its unparalleled adaptability and production speed. The Pfizer-BioNTech (BNT162b2) and Moderna (mRNA-1273) vaccines became the first mRNA vaccines authorized for emergency use, setting a precedent for future vaccine development16.
Several factors facilitated the rapid development of COVID-19 mRNA vaccines. First, researchers quickly sequenced the SARS-CoV-2 genome, identifying the spike protein as a key antigenic target. Synthetic mRNA encoding the spike protein was designed, optimized, and encapsulated in LNPs for delivery3. This approach bypassed the need for cell-based antigen production, reducing manufacturing timelines to mere weeks. Second, pre-existing platforms for mRNA synthesis and LNP formulation accelerated the transition from preclinical studies to clinical trials10.
Efficacy, Safety, and Adaptability
Efficacy and safety were key metrics in the success of COVID-19 mRNA vaccines. Clinical trials revealed that Pfizer-BioNTech and Moderna vaccines achieved efficacies of approximately 95% and 94%, respectively, in preventing symptomatic COVID-19 (Phase 3)17,18. These vaccines demonstrated strong safety profiles, with side effects generally limited to mild, self-limiting reactions such as fatigue and injection site pain. However, rare adverse events, including myocarditis and anaphylaxis, highlighted the need for continued monitoring and post-marketing surveillance11.
The adaptability of mRNA technology also proved instrumental in addressing SARS-CoV-2 variants. By modifying the mRNA sequence to match emerging viral mutations, manufacturers rapidly developed booster vaccines tailored to variant strains. This agility underscored the platform’s potential to combat evolving pathogens and laid the groundwork for its application to other viral diseases.
Future Outbreaks: Scalability and Multiplex Designs
Beyond COVID-19, the scalability and speed of mRNA vaccine development have implications for future outbreaks. The ability to design vaccines within days of identifying a pathogen’s genome could transform global responses to infectious diseases. Furthermore, mRNA platforms enable multiplexed vaccine designs, potentially targeting multiple pathogens or variants in a single formulation, enhancing immunization strategies.
The success of mRNA vaccines during the COVID-19 pandemic was a culmination of decades of research. Key advancements, such as incorporating pseudouridine into mRNA to improve stability and reduce immune activation, played a pivotal role12. Similarly, developing LNPs revolutionized vaccine delivery, ensuring that the fragile mRNA molecules could reach their target cells intact. These innovations enabled the rapid production of COVID-19 vaccines and provided a template for addressing future viral threats.
Expanding Applications of mRNA Technology
Building on the success during the COVID-19 pandemic, the potential of mRNA technology extends far beyond SARS-CoV-2. The lessons learned from COVID-19 vaccine development are now being applied to other infectious diseases, including influenza, RSV, and HIV. Researchers are exploring the use of mRNA vaccines to target highly mutable viruses, leveraging the platform’s flexibility to address antigenic drift and shift11. Additionally, mRNA technology is being investigated for its potential in cancer immunotherapy, which could encode tumor-specific antigens and stimulate robust anti-tumor immune responses.
In conclusion, the role of mRNA technology in COVID-19 vaccine development marked a turning point in medical science. Its ability to enable efficient vaccine production transformed the global response to the pandemic and established a foundation for future innovation. As researchers continue to refine mRNA platforms and address existing challenges, the potential to revolutionize healthcare through mRNA technology remains immense.
Advantages of mRNA Technology in Vaccine Development
Messenger RNA (mRNA) Technology has emerged as a transformative tool in vaccinology, offering numerous advantages over traditional approaches. Beyond its successful application in COVID-19 vaccines, mRNA technology is paving the way for innovative solutions against other viral diseases such as influenza and respiratory syncytial virus (RSV). Its unique properties enable rapid development, enhanced safety, high efficacy, and scalability, making it a cornerstone for future vaccine development.
Rapid Development and Production
One of the standout advantages of mRNA vaccines is their rapid development timeline. Traditional vaccine production often involves cultivating viruses in eggs or cell cultures, inactivating or attenuating them, and subsequently testing for safety and efficacy, a process that can take several years. In contrast mRNA vaccines can be designed and manufactured in weeks once the genetic sequence of a pathogen is known.
For example, the adaptability of mRNA technology allowed pharmaceutical companies to create updated vaccine candidates for COVID-19 variants, such as Omicron, within a matter of months. This speed is equally applicable to seasonal influenza vaccines, where matching circulating strains quickly is vital. Companies like Moderna and Pfizer are leveraging this technology to produce mRNA-based influenza vaccines that can adapt to new strains, potentially reducing the annual burden of flu-related illness19.
Enhanced Safety Profile
The mechanism of action for mRNA vaccines also contributes to their improved safety profile. Unlike traditional vaccines that may use live or inactivated viruses, mRNA vaccines rely on delivering a genetic blueprint for cells to produce antigens. This eliminates the risk of vaccine-induced infections, a concern particularly relevant in immunocompromised populations. Furthermore, mRNA does not integrate into the host genome, as it is inherently unstable and degrades naturally after delivering its message. This transient presence in the body minimizes the potential for long-term adverse effects, enhancing public confidence in vaccine safety20.
High Efficacy and Immunogenicity
mRNA vaccines are engineered to closely mimic the natural structure of viral proteins, enabling them to induce robust immune responses. This fidelity ensures that the immune system effectively recognizes and targets the pathogen, translating into high efficacy rates. For instance, studies on mRNA vaccines for RSV and influenza have demonstrated their ability to produce strong neutralizing antibody responses, critical for preventing severe disease. Preclinical trials have also highlighted the potential of mRNA vaccines to protect against emerging viruses such as avian influenza (H5N1) and even zoonotic diseases21.
Moreover, mRNA technology facilitates the inclusion of helper molecules, such as LNPs, which protect the fragile RNA and enhance its delivery into cells. These LNPs also act as adjuvants, further boosting the vaccine’s immunogenicity. This dual function significantly reduces the need for additional adjuvants, streamlining vaccine design.
Versatility and Scalability
The scalability and versatility of mRNA vaccine technology represent major advancements over traditional vaccine platforms. Unlike egg-based or cell-culture systems, which are time-intensive and vulnerable to supply chain disruptions, mRNA synthesis relies on in vitro transcription, enabling rapid, large-scale production independent of live pathogens11,22. This capability was critical during the COVID-19 pandemic, allowing manufacturers to produce billions of doses in record time, demonstrating the efficiency of standardized mRNA manufacturing19. Beyond scalability, the modular nature of mRNA technology allows for quick adaptation to different pathogens by altering the encoded antigen. This adaptability is particularly valuable for addressing diseases with high mutation rates, such as influenza and HIV. For example, Moderna is leveraging mRNA to develop broadly neutralizing antibodies against HIV23. These advantages position mRNA vaccines as a transformative tool for both pandemic preparedness and targeted immunotherapies.
Multivalent and Personalized Vaccine Potential
Inactivated and live‑attenuated influenza vaccines are inexpensive and established but require 6–9 months of egg‑based production, limiting agility when strains drift24. Adenoviral‑vector vaccines can elicit strong cellular immunity but are constrained by pre‑existing anti‑vector immunity that may blunt boosters16. Protein‑subunit approaches are highly safe yet often depend on potent adjuvants for robust T‑cell responses6. By contrast, mRNA platforms pair rapid design cycles with strong humoral and cellular immunity, albeit with current cold‑chain and cost constraints.
mRNA technology enables the development of multivalent vaccines, which target multiple strains or variants of a virus within a single formulation. This capability is especially significant for viruses like influenza, which require vaccines that cover several subtypes. A multivalent mRNA influenza vaccine could provide broader protection and reduce the need for annual reformulations.
Additionally, mRNA technology holds promise for personalized medicine. Researchers are exploring its application in cancer vaccines, where mRNA can encode tumor-specific antigens to stimulate an immune response tailored to an individual’s unique cancer profile. This personalized approach has implications for infectious diseases as well, where mRNA vaccines could be customized to target specific strains circulating in different regions25.
Challenges and Limitations of mRNA Vaccines
While messenger RNA (mRNA) technology has revolutionized vaccine development, especially with the success of COVID-19 vaccines, it is not without its challenges and limitations. Addressing these issues is critical for the broader applications of mRNA vaccines to other viral diseases, such as influenza and respiratory syncytial virus (RSV), and for realizing their full potential in global healthcare.
Stability and Storage Requirements
Despite the success of mRNA vaccines, their stability and stringent cold storage requirements present significant challenges, particularly in low-resource settings. mRNA is inherently unstable and highly susceptible to degradation by ribonucleases, necessitating ultra-cold storage conditions to maintain efficacy. For instance, the Pfizer–BioNTech COVID-19 vaccine initially required storage at -80°C to -60°C, although later formulations improved stability to allow refrigeration3. These logistical hurdles have limited vaccine accessibility in low-income regions, underscoring the need for thermostable formulations. Efforts are underway to develop lyophilized (freeze-dried) mRNA vaccines and novel excipient-stabilized formulations that could enable storage at standard refrigeration temperatures, potentially improving global distribution and equity26.
Delivery Efficiency
Delivering mRNA into cells effectively and safely poses another technical challenge. mRNA molecules are large and negatively charged, making it difficult for them to cross cell membranes. LNPs have emerged as an efficient delivery system, encapsulating mRNA to facilitate cellular uptake while protecting it from degradation. However, LNPs are complex to produce, requiring advanced manufacturing techniques and stringent quality controls. Scaling up production of these nanoparticles to meet global demands introduces logistical and cost challenges, particularly in resource-limited regions. Moreover, while LNPs enhance delivery, they are associated with some individuals’ reactogenicity (e.g., injection site pain and transient inflammation), which can impact vaccine acceptance27.
Immunogenicity and Reactogenicity
In perspective, systemic adverse events are broadly comparable to those following inactivated influenza vaccines and far lower than complications from influenza infection itself, which drives substantial annual hospitalizations28. Myocarditis/pericarditis after mRNA vaccination is rare—on the order of tens per million second doses in younger males—and remains less frequent than myocarditis following SARS‑CoV‑2 infection29.
The robust immune responses elicited by mRNA vaccines are both an advantage and a limitation. While these vaccines produce strong neutralizing antibody responses, some recipients experience mild to moderate side effects, including fever, fatigue, and headaches. These effects are attributed to the activation of innate immune pathways by LNPs or the mRNA itself, which can sometimes lead to excessive inflammation. For example, a study noted that certain components of LNPs can trigger inflammatory responses, highlighting the need for optimization in delivery systems to mitigate such effects. Further research into refining the composition of LNPs and mRNA constructs could enhance vaccine tolerability while maintaining efficacy30.
Manufacturing Complexity and Cost
Quality control of IVT RNA, capping, and LNP assembly adds cost layers relative to egg‑based influenza vaccines; analysts project mRNA seasonal flu vaccines could be several‑fold more expensive than conventional shots without scale efficiencies31. Batch‑to‑batch consistency requires tight control of transcription yield, 5′‑capping, and nanoparticle size distributions; regulators mandate orthogonal analytics to ensure potency and purity across lots14.
Despite claims of scalability, the production of mRNA vaccines involves complex processes that require advanced infrastructure and highly specialized materials. Unlike traditional vaccines, mRNA vaccines demand cutting-edge facilities for synthesizing RNA and formulating LNPs. Quality control during large-scale production is crucial to ensure consistency in efficacy and safety, which adds to manufacturing costs. Although the speed of mRNA vaccine development may offset these costs compared to conventional approaches, the financial burden could limit accessibility, particularly in low- and middle-income countries32.
Public Perception and Misinformation
Overall, current evidence indicates cost and manufacturing complexity are the most immediate barriers to routine mRNA use for influenza and RSV, whereas cold‑chain limits are being mitigated by advances in thermostability and lyophilization33,34. The rapid development of mRNA vaccines during the COVID-19 pandemic led to unprecedented scientific breakthroughs, but it also gave rise to skepticism and misinformation. Concerns about the long-term safety of mRNA vaccines, their speed of development, and misunderstandings about how they work have contributed to vaccine hesitancy. Although studies confirm the safety and efficacy of mRNA vaccines, addressing public concerns through transparent communication and education is essential to foster trust and promote widespread acceptance35.
Targeting Complex Pathogens
While mRNA technology has shown remarkable success against COVID-19, extending its application to other diseases presents unique challenges. Viral pathogens such as RSV and influenza are highly variable, with rapidly mutating strains that complicate vaccine design. Developing a multivalent mRNA vaccine to provide broad protection against these pathogens requires extensive research to identify conserved epitopes and optimize antigen selection. Additionally, achieving durable immunity against pathogens with immune-evasion mechanisms, such as HIV, remains a significant scientific hurdle36.
Regulatory and Ethical Considerations
For routine indications such as seasonal influenza or pediatric RSV, regulators require full Phase 3 evidence, larger safety databases, and longer follow‑up (often ≥12–24 months), followed by post‑licensure pharmacovigilance—distinct from pandemic EUAs37,38. Global equity efforts, including the WHO mRNA Technology Transfer Hub, aim to decentralize manufacturing capacity to reduce cost and access barriers34.
The regulatory framework for mRNA vaccines is still evolving. Emergency-use authorizations, like those granted during the COVID-19 pandemic, allowed rapid deployment but may not always apply to vaccines for other diseases. Long-term safety data for mRNA technology is still being collected, and regulators must balance speed with comprehensive efficacy and safety evaluations. Furthermore, ensuring equitable distribution of these vaccines globally is an ethical challenge that requires international collaboration and policy reform34.
Expansion of mRNA Vaccines Beyond COVID-19
The successful application of mRNA technology during the COVID-19 pandemic has revolutionized vaccine development, offering a fast, flexible, and scalable platform. This means, mRNA vaccines have a growing potential to combat a wide range of viral diseases beyond COVID-19. By targeting both well-known and emerging pathogens mRNA vaccines hold promise for addressing significant unmet needs in global health. The rest of this section will be divided into subsections, each highlighting a specific disease, detailing its challenges, and the innovative ways mRNA technology is utilized to develop effective vaccines.
Influenza
Influenza remains a persistent global health challenge, causing significant illnesses of about 9.3 million to 41 million annually28. Conventional influenza vaccines are developed based on predictions of circulating strains, often leading to mismatches between vaccine composition and emerging strains. This results in variable efficacy, averaging 40–60% in most years. The rapid mutation of influenza viruses (antigenic drift and shift) underscores the need for a more adaptable vaccine platform28.
mRNA technology offers transformative solutions to these challenges. By enabling the rapid synthesis of mRNA sequences encoding hemagglutinin (HA) or neuraminidase (NA) antigens, mRNA vaccines can be designed to match newly identified influenza strains in record time. Moderna and Pfizer have initiated clinical trials for mRNA-based influenza vaccines. Early-phase results suggest that these vaccines elicit robust antibody responses, comparable to or surpassing traditional egg-based vaccines, with added versatility to include multiple antigenic targets in a single formulation39.
Furthermore, the modularity of mRNA technology facilitates the incorporation of conserved influenza epitopes, which could enhance cross-strain protection and mitigate the impact of antigenic drift. This approach aims to create a universal influenza vaccine, a longstanding goal in vaccinology, by targeting conserved regions like the HA stalk domain. Recent research suggests that combining HA stalk antigens with nanoparticle delivery systems could enhance efficacy by stabilizing antigenic structures24.
Recent advancements in mRNA-based influenza vaccines have demonstrated promising results. A Phase 1/2 clinical trial evaluating Moderna’s investigational quadrivalent influenza vaccine, mRNA-1010, reported that a single dose elicited robust hemagglutination inhibition (HAI) titers that persisted through six months. The vaccine was generally well-tolerated, with no vaccine-related serious adverse events or deaths reported30. Additionally, GSK announced positive headline results from a Phase II trial for its mRNA seasonal influenza vaccine program, indicating that the vaccine candidates elicited strong overall antibody titers with an acceptable safety profile in both younger and older adults40.
In addition to their immunogenic advantages, mRNA vaccines like mRNA-1010 offer significant production scalability and cost-effectiveness benefits. The cell-free manufacturing process of mRNA vaccines allows for rapid and scalable production, with estimates suggesting that a 5-liter bioreactor can produce nearly a million doses in a single batch31. This efficiency accelerates vaccine availability and reduces production costs compared to traditional vaccine platforms. Financial projections indicate that mRNA-1010 could generate annual revenues of approximately $407 million globally by 203841.
However, a notable challenge for mRNA-1010 is its thermostability. Like many mRNA vaccines, it requires storage at subzero temperatures to maintain stability and efficacy, which can complicate distribution, especially in regions lacking robust cold chain infrastructure. During high-pressure situations such as pandemics, this requirement can lead to significant vaccine wastage if proper storage conditions are not maintained. For instance, during the COVID-19 pandemic, the necessity for ultra-cold storage resulted in vaccine spoilage in areas without adequate refrigeration facilities, and this could happen to mRNA-1010 as well. Nonetheless, ongoing research aims to develop lyophilized (freeze-dried) mRNA lipid nanoparticles to enhance stability at room temperature, thereby simplifying storage and transportation logistics33.
Overall, mRNA-based influenza vaccines represent a significant leap forward in combating the ever-evolving influenza virus. Their rapid adaptability, robust immunogenicity, and potential for broad-spectrum protection position them as a promising alternative to traditional egg-based vaccines. As clinical trials continue to refine formulation strategies, solve delivery challenges, and evaluate long-term efficacy, mRNA could pave the way for a more effective and universal influenza vaccine. With the ability to swiftly respond to emerging strains and incorporate conserved antigenic targets, mRNA vaccines hold the potential to revolutionize seasonal influenza prevention and enhance global public health resilience.
Respiratory Syncytial Virus (RSV)
Respiratory Syncytial Virus (RSV) is a leading cause of severe respiratory infections in infants, the elderly, and immunocompromised individuals. Despite decades of research, no effective RSV vaccine has been widely adopted. The challenges stem from RSV’s ability to evade immune detection and difficulty eliciting durable immunity against the virus6.
mRNA vaccines are uniquely positioned to overcome these obstacles. By encoding stabilized prefusion forms of the RSV F protein—a key antigen for neutralizing antibodies—mRNA vaccines can induce strong and targeted immune responses. Some vaccine development companies have reported promising clinical trial data for RSV mRNA vaccines, with robust immunogenicity and safety profiles in both elderly adults and pediatric populations. For instance, Moderna’s investigational RSV vaccine, mRNA-1345, demonstrated vaccine efficacy of 83.7% against RSV-associated lower respiratory tract disease with at least two symptoms in older adults. The vaccine was generally well-tolerated, with most adverse reactions being mild to moderate in severity and transient39,42,43.
Furthermore, a Phase 1 clinical trial evaluating the safety and immunogenicity of mRNA-1345 reported that the vaccine was well-tolerated and immunogenic following a single injection and a 12-month booster. The most frequently reported solicited adverse reactions were injection site pain, fatigue, headache, arthralgia, and myalgia, which were mostly mild to moderate in severity and transient. A single mRNA-1345 injection boosted RSV-A and RSV-B neutralizing antibody titers and prefusion F binding antibody concentrations at 1 month, with geometric mean fold rises ranging from 10.2 to 16.5 for RSV-A and 5.3 to 12.5 for RSV-B. RSV antibody levels remained above baseline through 12 months, indicating immune persistence. A 12-month booster injection also increased RSV-A and RSV-B neutralizing antibody titers and prefusion F binding antibody concentrations; titers after the booster injection were numerically lower than those after the first dose, with overlapping 95% confidence intervals7.
Lastly, combining RSV antigens with other respiratory virus targets in a single multivalent mRNA vaccine can also be explored. Such approaches could streamline immunization schedules and improve vaccine coverage across vulnerable populations. Recent innovations in LNP formulations also enhance RSV vaccine delivery, reducing systemic inflammation while maintaining efficacy44. These findings underscore the potential of mRNA-based vaccines in providing effective protection against RSV, particularly in populations at higher risk for severe disease.
All in all, mRNA vaccine technology’s adaptability and rapid development cycle have opened new avenues for combating various viral diseases beyond COVID-19. Researchers are actively exploring mRNA vaccine candidates for pathogens such as HIV, Zika virus, rabies, Ebola virus, cytomegalovirus (CMV), dengue virus, and hepatitis C virus (HCV). The ability to swiftly design and produce mRNA vaccines tailored to specific pathogens holds promise for addressing these significant global health challenges. As research progresses, mRNA vaccines may become integral tools in preventing a broad spectrum of infectious diseases, improving public health outcomes worldwide.
Future Directions for mRNA Vaccine Development
Messenger RNA (mRNA) technology has fundamentally reshaped the field of vaccinology, establishing a framework of rapid, scalable, and highly adaptable vaccine development. While its successful application during the COVID-19 pandemic demonstrated its transformative potential, the future of mRNA technology lies in addressing existing barriers, optimizing the platform, and expanding its utility to combat a broader spectrum of viral diseases. By overcoming current limitations and integrating novel advancements, mRNA technology could redefine how the world addresses public health threats.
Developing Universal Vaccines
The flexibility of mRNA technology enables the design of vaccines targeting conserved epitopes across multiple viral strains, paving the way for universal vaccines. For example, researchers are leveraging mRNA platforms to develop a universal influenza vaccine. By encoding conserved regions of the hemagglutinin (HA) protein, particularly the stalk domain, mRNA vaccines could provide cross-strain protection against seasonal and pandemic influenza strains39. This concept is being extended to other viral pathogens, including respiratory syncytial virus (RSV) and HIV, where conserved antigenic sites are targeted to elicit broad and durable immunity45.
Additionally, advances in bioinformatics and machine learning are aiding in identifying highly conserved epitopes. These computational tools can analyze viral genomes to predict potential targets for mRNA vaccines, accelerating the design process. For instance, algorithms are employed to identify conserved sequences in emerging zoonotic viruses, providing a head start in vaccine development for future pandemics46.
Addressing Antigenic Diversity and Immune Evasion
For viruses with high mutation rates, such as HIV and hepatitis C virus (HCV), antigenic diversity poses a significant challenge. mRNA technology offers the flexibility to encode multiple antigens within a single vaccine formulation, potentially overcoming this issue. For instance, multivalent mRNA vaccines can include genetic instructions for producing various viral proteins, offering a broader protection45. This strategy is particularly relevant for HIV, where mRNA vaccines are designed to deliver sequential immunogens that guide the immune system toward producing broadly neutralizing antibodies45.
Another innovative approach involves the use of epitope-focused vaccines. By engineering mRNA to encode only the most immunogenic and conserved portions of viral proteins, researchers aim to enhance vaccine efficacy against diverse viral strains23,21. Such epitope-focused designs are under active investigation for HIV, HCV, and dengue virus, where antigenic variability has historically hindered vaccine development46.
Combining Therapeutics with Vaccines
mRNA technology’s potential extends beyond prophylactic vaccines to therapeutic applications. Researchers are exploring its use in combination vaccines that deliver therapeutic agents alongside immunogens. For example, mRNA vaccines encoding cytokines or immune checkpoint inhibitors could boost the immune response against viruses that establish chronic infections, such as hepatitis B virus (HBV) and Epstein-Barr virus17,47. This dual approach could also be applied to oncogenic viruses, such as human papillomavirus, to prevent infection and target virus-induced tumors simultaneously48.
Additionally, preclinical studies combine mRNA vaccines with monoclonal antibody therapies. By encoding instructions for the host to produce therapeutic antibodies, mRNA vaccines could offer an alternative to traditional antibody treatments, which are expensive and require complex manufacturing processes3. This approach is particularly promising for emerging zoonotic viruses and diseases with limited therapeutic options47.
Expanding Global Vaccine Manufacturing and Equity
The scalability of mRNA vaccine production is a significant advantage, but disparities in global manufacturing capacity remain a barrier to equitable vaccine distribution. Establishing decentralized production hubs in low- and middle-income countries could mitigate this issue. Initiatives like the WHO’s mRNA Technology Transfer Hub aim to equip developing nations with the knowledge and infrastructure needed to produce mRNA vaccines locally. These efforts are bolstered by developing portable mRNA production units, such as biofoundries, which could rapidly produce vaccines in outbreak scenarios46.
Collaborative efforts between academic institutions, biotech companies, and governments will be critical to ensuring equitable access to mRNA vaccines. Policies promoting technology sharing, open-source platforms, and tiered pricing structures could further democratize access. In addition, integrating mRNA vaccine production with existing immunization programs could streamline the rollout of new vaccines for endemic diseases39.
Exploring Personalized Vaccinology
Personalized medicine represents the frontier of mRNA vaccine technology. By tailoring vaccines to an individual’s genetic and immunological profile, researchers aim to maximize efficacy and minimize adverse effects. For example, personalized mRNA vaccines are developed for caner immunotherapy, encoding tumor-specific antigens based on a patient’s unique mutational landscape45. Similar principles could be applied to infectious diseases, where regional or individual variations in pathogen strains necessitate customized vaccine formulations46.
The use of advanced genomic sequencing and bioinformatics is central to this effort. Researchers can design mRNA vaccines that address localized outbreaks or emerging variants by analyzing pathogen genomes in real-time. This approach can potentially revolutionize outbreak management, enabling targeted interventions that reduce the spread of disease46.
Conclusion
The advent of mRNA technology has profoundly transformed the landscape of vaccine development, establishing a new paradigm for rapid, adaptable, and scalable vaccine platforms. Its revolutionary impact was demonstrated during the COVID-19 pandemic, where mRNA vaccines developed by Pfizer-BioNTech and Moderna were created, tested, and distributed in record time. These vaccines showcased high efficacy and robust safety profiles and emphasized the flexibility of mRNA technology to address emerging viral variants through rapid updates. This success has positioned mRNA as a cornerstone for pandemic preparedness, setting a precedent for developing vaccines against other viral diseases.
Beyond COVID-19, mRNA technology holds immense promise for tackling long-standing challenges in vaccinology. Its ability to encode multiple antigens allows for the development of multivalent vaccines, addressing highly mutable viruses like influenza, respiratory syncytial virus (RSV), and HIV. The modular design of mRNA vaccines enables rapid adaptation to new pathogens, overcoming the limitations of traditional vaccine platforms that often require years to develop and manufacture. Additionally, the potential to incorporate conserved epitopes into mRNA vaccines offers hope for achieving cross-strain protection, paving the way for universal vaccines that target diseases with high antigenic variability.
Despite its transformative potential, several challenges must be addressed to maximize the impact of mRNA vaccines. Current storage requirements, often demanding ultra-cold temperatures, limit accessibility in resource-constrained regions. Efforts to develop thermostable formulations could significantly enhance global vaccine distribution. Similarly, refining delivery systems, such as lipid nanoparticles, is essential to improve efficiency, reduce side effects, and lower production costs. Moreover, addressing public skepticism and misinformation about mRNA technology will be critical to ensuring widespread acceptance and trust.
The broader implications of mRNA technology extend beyond infectious diseases, offering innovative solutions for cancer immunotherapy and other therapeutic applications. Personalized mRNA vaccines tailored to an individual’s genetic and immunological profile are already under investigation, highlighting the potential for precision medicine. Furthermore, integrating mRNA platforms into global health initiatives could revolutionize responses to emerging diseases, reduce inequities in vaccine access, and provide cost-effective solutions for endemic illnesses.
In conclusion, mRNA technology represents a groundbreaking advancement in vaccine development, with the capacity to combat both emerging and persistent viral threats. Continued investment in research and development is vital to overcome existing limitations and expand its applications. As researchers refine the platform and address logistical and technical challenges, mRNA vaccines are poised to redefine global healthcare, offering unprecedented opportunities to safeguard public health in the face of evolving diseases.
References
- Worldometer. (2024, April 13). Coronavirus cases:. Worldometer. https://www.worldometers.info/coronavirus/. [↩]
- World Health Organization. (n.d.). Covid-19 cases | WHO COVID-19 Dashboard. World Health Organization. https://data.who.int/dashboards/covid19/cases. [↩]
- Polack, F. P., et al (2020). Safety and efficacy of the BNT162B2 mrna covid-19 vaccine. New England Journal of Medicine, 383(27), 1576–1578. https://doi.org/10.1056/NEJMoa2034577. [↩] [↩] [↩] [↩]
- Baden, L. R., et al. “Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine.” The New England Journal of Medicine, vol. 384, no. 5, 2021, pp. 403–416. https://doi.org/10.1056/NEJMoa2035389. [↩]
- CDC. (2024, November 13). About estimated flu burden. Centers for Disease Control and Prevention. https://www.cdc.gov/flu-burden/php/about/index.html. [↩]
- Roberts, J. N., Graham, B. S., Karron, R. A., Munoz, F. M., Falsey, A. R., Anderson, L. J., Marshall, V., Kim, S., & Beeler, J. A. (2016). Challenges and opportunities in RSV Vaccine Development: Meeting Report from FDA/NIH Workshop. Vaccine, 34(41), 4843–4849. https://doi.org/10.1016/j.vaccine.2016.07.057. [↩] [↩] [↩]
- Shaw, C. A., Essink, B., Harper, C., Mithani, R., Kapoor, A., Dhar, R., Wilson, L., Guo, R., Panozzo, C. A., Wilson, E., Simorellis, A. K., Reuter, C., Stoszek, S. K., Chen, G. L., Das, R., & Goswami, J. (2024). Safety and immunogenicity of an mrna-based RSV vaccine including a 12-month booster in a phase 1 clinical trial in healthy older adults. The Journal of Infectious Diseases, 230(3). https://doi.org/10.1093/infdis/jiae081. [↩] [↩]
- Sahin, U., Karikó, K., & Türeci, Ö. (2014). mRNA-based therapeutics—developing a new class of drugs. Nature Reviews Drug Discovery, 13(10), 759–780. https://doi.org/10.1038/nrd4278. [↩] [↩]
- Chaudhary, N., Weissman, D., & Whitehead, K. A. (2021). mRNA vaccines for infectious diseases: Principles, delivery and clinical translation. Nature Reviews Drug Discovery, 20(11), 817–838. https://doi.org/10.1038/s41573-021-00283-5. [↩] [↩] [↩]
- Corbett, K.S., Edwards, D.K., Leist, S.R. et al (2020). SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature 586, 567–571. https://doi.org/10.1038/s41586-020-2622-0. [↩] [↩] [↩] [↩]
- Pardi, N., Hogan, M. J., Porter, F. W., & Weissman, D. et al (2018). mRNA vaccines — a new era in Vaccinology. Nature Reviews Drug Discovery, 17(4), 261–279. https://doi.org/10.1038/nrd.2017.243. [↩] [↩] [↩] [↩] [↩] [↩]
- Karikó, K., Muramatsu, H., Welsh, F. A., Ludwig, J., Kato, H., Akira, S., & Weissman, D. et al (2008). Incorporation of pseudouridine into mrna yields superior nonimmunogenic vector with increased translational capacity and biological stability. Molecular Therapy, 16(11), 1833–1840. https://doi.org/10.1038/mt.2008.200. [↩] [↩]
- Lundstrom, K. “Self-Replicating RNA Viruses for Vaccine Development against Infectious Diseases and Cancer.” Vaccines, vol. 6, no. 2, 2018, p. 28. https://doi.org/10.3390/vaccines6020028. [↩]
- Kwon, J., Lee, Y., Lee, H., & Shin, S. (2022). Advances in mRNA therapeutics: Systematic review of the manufacturing processes and challenges. Journal of Pharmaceutical Investigation, 52(1), 1–15. https://doi.org/10.1007/s40005-021-00539-0. [↩] [↩]
- Liu, M. A., VanBlargan, L. A., Rothlauf, P. W., & Bloom, J. D. (2023). Rapid adaptation of mRNA vaccines for emerging viral threats. Cell Reports Medicine, 4(3), 100926. https://doi.org/10.1016/j.xcrm.2023.100926. [↩]
- Cox, R. J., & Peacock, S. J. (2021). mRNA vaccines for SARS-CoV-2: The tipping point for vaccines and the future. NPJ Vaccines, 6, 1-8. https://doi.org/10.1038/s41541-021-00356-x. [↩] [↩]
- Polack, F. P., et al (2020). Safety and efficacy of the BNT162B2 mrna covid-19 vaccine. New England Journal of Medicine, 383(27), 1576–1578. https://doi.org/10.1056/NEJMoa2034577. [↩] [↩]
- Baden, L. R., et al. “Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine.” The New England Journal of Medicine, vol. 384, no. 5, 2021, pp. 403–416. https://doi.org/10.1056/NEJMoa2035389. [↩]
- Barton, C. (2024, August 7). MRNA technologies: Looking beyond covid vaccines. pharmaphorum. https://pharmaphorum.com/rd/mrna-technologies-looking-beyond-covid-vaccines. [↩] [↩]
- Zhang, C., Maruggi, G., Shan, H., & Li, J. (2019). Advances in mrna vaccines for infectious diseases. Frontiers in Immunology, 10. https://doi.org/10.3389/fimmu.2019.00594. [↩]
- Stobbe, M., & Neergaard, L. (2024, May 31). Scientists are testing mrna vaccines to protect cows and people against Bird Flu. AP News. https://apnews.com/article/bird-flu-mrna-vaccine-ab3cf2df5ef7dca8f56d279205a97ff7. [↩] [↩]
- U.S. National Library of Medicine. (n.d.). What are mrna vaccines and how do they work?: Medlineplus Genetics. MedlinePlus. https://medlineplus.gov/genetics/understanding/therapy/mrnavaccines/. [↩]
- Laise, E. (2024, September 4). Moderna’s experimental mpox vaccine reduces severity of disease, early study finds. MarketWatch. https://www.marketwatch.com/story/modernas-experimental-mpox-vaccine-reduces-severity-of-disease-early-study-finds-36ff0b7a. [↩] [↩]
- Krammer, F., & Palese, P. (2015). Advances in the development of influenza virus vaccines. Nature Reviews Drug Discovery, 14(3), 167–182. https://doi.org/10.1038/nrd4529. [↩] [↩]
- Mole, B. (2024, July 4). US government awards moderna $176 million for mrna bird flu vaccine. Wired. https://www.wired.com/story/moderna-mrna-vaccine-bird-flu-h5n1/. [↩]
- U.S. National Library of Medicine. (n.d.). PubMed. National Center for Biotechnology Information. https://pubmed.ncbi.nlm.nih.gov/. [↩]
- Lipsitch, M., & Kahn, R. (2021). Interpreting vaccine efficacy trial results for infection and transmission. MedRXiv. https://doi.org/10.1101/2021.02.25.21252415. [↩]
- CDC. (2024, November 13). About estimated flu burden. Centers for Disease Control and Prevention. https://www.cdc.gov/flu-burden/php/about/index.html. [↩] [↩] [↩]
- Mevorach, D., et al. “Myocarditis after BNT162b2 mRNA Vaccine against Covid-19 in Israel.” The New England Journal of Medicine, vol. 385, no. 23, 2021, pp. 2140–2149. https://doi.org/10.1056/NEJMoa2109730. [↩]
- Lee, J., Woodruff, M. C., Kim, E. H., & Nam, J.-H. (2023). Knife’s Edge: Balancing immunogenicity and reactogenicity in mrna vaccines. Experimental & Molecular Medicine, 55(7), 1305–1313. https://doi.org/10.1038/s12276-023-00999-x. [↩] [↩]
- Plotkin, S., Robinson, J. M., Cunningham, G., Iqbal, R., & Larsen, S. (2017). The complexity and cost of Vaccine Manufacturing – An Overview. Vaccine, 35(33), 4064–4071. https://doi.org/10.1016/j.vaccine.2017.06.003. [↩] [↩]
- Explore scientific, technical, and medical research on ScienceDirect. ScienceDirect.com | Science, health and medical journals, full text articles and books. (n.d.). https://www.sciencedirect.com/. [↩]
- Uddin, M. N., & Roni, M. A. (2021). Challenges of storage and stability of mrna-based COVID-19 vaccines. Vaccines, 9(9), 1033. https://doi.org/10.3390/vaccines9091033. [↩] [↩]
- World Health Organization. (2021). Vaccine equity. World Health Organization. https://www.who.int/campaigns/vaccine-equity?utm_source. [↩] [↩] [↩]
- Miles, T. (2019, May 21). Vaccine doubts spread like disease, must be taken offline: Vaccine chief . Reuters. https://www.reuters.com/article/us-health-vaccines/vaccine-doubts-spread-like-disease-must-be-taken-offline-vaccine-chief-idUSKCN1SR1SV/. [↩]
- Kumar, A., Blum, J., Thanh Le, T., Havelange, N., Magini, D., & Yoon, I.-K. (2022). The mrna vaccine development landscape for infectious diseases. Nature Reviews Drug Discovery, 21(5), 333–334. https://doi.org/10.1038/d41573-022-00035-z. [↩]
- U.S. Food and Drug Administration. “Emergency Use Authorization for Vaccines to Prevent COVID-19: Guidance for Industry.” 2022. https://www.fda.gov/. [↩]
- European Medicines Agency. “Guideline on Clinical Evaluation of Vaccines.” 2023. https://www.ema.europa.eu/. [↩]
- Moderna announces interim phase 3 safety and immunogenicity results for mrna-1010, a seasonal influenza vaccine candidate. Moderna. (2023, February 16). https://investors.modernatx.com/news/news-details/2023/Moderna-Announces-Interim-Phase-3-Safety-and-Immunogenicity-Results-for-mRNA-1010-a-Seasonal-Influenza-Vaccine-Candidate/default.aspx. [↩] [↩] [↩] [↩]
- GSK. (2024). GSK’s mRNA-based influenza vaccine demonstrates positive results in Phase II trial. Retrieved from https://www.gsk.com/en-gb/media/press-releases/gsk-s-mrna-based-influenza-vaccine-demonstrates-positive-results-in-phase-ii-trial/. [↩]
- Kgi-Admin. (2024, August 27). Risk adjusted net present value: What is the current valuation of Moderna’s mrna-1010. Pharmaceutical Technology. https://www.pharmaceutical-technology.com/data-insights/mrna-1010-moderna-net-present-value/. [↩]
- Sullender, W. (2000). Respiratory syncytial virus genetic and antigenic diversity. Clinical Microbiology Reviews, 13(1), 1–15. https://doi.org/10.1128/cmr.13.1.1-15.2000. [↩]
- Topalidou, X., Kalergis, A. M., & Papazisis, G. (2023). Respiratory syncytial virus vaccines: A review of the candidates and the approved vaccines. Pathogens, 12(10), 1259. https://doi.org/10.3390/pathogens12101259. [↩]
- Espeseth, A. S., Cejas, P. J., Citron, M. P., Wang, D., DiStefano, D. J., Callahan, C., Donnell, G. O., Galli, J. D., Swoyer, R., Touch, S., Wen, Z., Antonello, J., Zhang, L., Flynn, J. A., Cox, K. S., Freed, D. C., Vora, K. A., Bahl, K., Latham, A. H., … Bett, A. J. (2020). Modified mrna/lipid nanoparticle-based vaccines expressing respiratory syncytial virus F protein variants are immunogenic and protective in rodent models of RSV infection. Npj Vaccines, 5(1). https://doi.org/10.1038/s41541-020-0163-z. [↩]
- Barouch, D. H. (2008). Challenges in the development of an HIV-1 vaccine. Nature, 455(7213), 613–619. https://doi.org/10.1038/nature07352. [↩] [↩] [↩] [↩]
- World Health Organization. (n.d.). Covid-19 cases | WHO COVID-19 Dashboard. World Health Organization. https://data.who.int/dashboards/covid19/cases. [↩] [↩] [↩] [↩] [↩]
- Rappuoli, R., Bottomley, M. J., D’Oro, U., Finco, O., & De Gregorio, E. (2016). Reverse Vaccinology 2.0: Human immunology instructs vaccine antigen design. Journal of Experimental Medicine, 213(4), 469–481. https://doi.org/10.1084/jem.20151960. [↩] [↩]
- Pfizer and BioNTech Confirm High Efficacy and No Serious Safety Concerns Through Up to Six Months Following Second Dose in Updated Topline Analysis of Landmark COVID-19 Vaccine Study. Pfizer. (2021, April 1). https://www.pfizer.com/news/press-release/press-release-detail/pfizer-and-biontech-confirm-high-efficacy-and-no-serious. [↩]




